EP2170372A1 - Compositions de stérol 24-c-méthyltransférase de leishmania pour la prévention, le traitement et le diagnostic de leishmanioses - Google Patents

Compositions de stérol 24-c-méthyltransférase de leishmania pour la prévention, le traitement et le diagnostic de leishmanioses

Info

Publication number
EP2170372A1
EP2170372A1 EP08781725A EP08781725A EP2170372A1 EP 2170372 A1 EP2170372 A1 EP 2170372A1 EP 08781725 A EP08781725 A EP 08781725A EP 08781725 A EP08781725 A EP 08781725A EP 2170372 A1 EP2170372 A1 EP 2170372A1
Authority
EP
European Patent Office
Prior art keywords
polypeptide
smt
cells
composition
leishmania
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP08781725A
Other languages
German (de)
English (en)
Inventor
Steven Reed
Yasuyuki Goto
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Access To Advanced Health Institute
Original Assignee
Infectious Disease Research Institute Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Infectious Disease Research Institute Inc filed Critical Infectious Disease Research Institute Inc
Publication of EP2170372A1 publication Critical patent/EP2170372A1/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/45Transferases (2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/002Protozoa antigens
    • A61K39/008Leishmania antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55572Lipopolysaccharides; Lipid A; Monophosphoryl lipid A
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates generally to compositions and methods for preventing, treating and detecting leishmaniasis in patients. More particularly, the invention relates to compositions comprising Leishmania sterol 24-c-methyltransferase
  • SMT polypeptides, fusion polypeptides thereof, as well as polynucleotides encoding such polypeptides and fusions.
  • Human leishmaniasis is a spectrum of diseases caused by protozoan parasites of the genus Leishmania. Leishmaniases are roughly classified into three types of diseases, cutaneous leishmaniasis (CL), mucosal leishmaniasis (ML) and visceral leishmaniasis (VL), according to the clinical manifestations. Visceral leishmaniasis, generally caused by species of the L. donovani complex, i.e., L. donovani and L. infantum (chagasi), is the most severe form, with approximately 500,000 new cases reported annually (information from World Health Organization: www.who.int/leishmaniasis/en/).
  • Active VL is characterized by hematological and hepatosplenic abnormalities, and is generally fatal unless properly treated.
  • Leishmania parasites are transmitted by the bite of sandflies and the infecting promastigotes differentiate into and replicate as amastigotes within macrophages in the mammalian host.
  • cellular immune responses are critical for protection against leishmaniasis.
  • ThI immune responses play an important role in mediating protection against Leishmania, including roles for CD4 + and CD8 + T cells, IFN- ⁇ , IL- 12, TNF- ⁇ and NO, whereas inhibitory effects have been reported for IL-IO and TGF- ⁇ (Engwerda et al., (1998) Eur J Immunol 28:669-680; Murphy et al., (2001) Eur J Immunol 31 :2848-2856; Murray and Nathan. (1999) J Exp Med 189:741-746; Murray et al., (2000) Infect Immun 68:6289-6293; Squires et al., (1989) J Immunol 143:4244-4249; Taylor and Murray.
  • Immunization against leishmaniasis in animal models can be effected by delivery of antigen-encoding DNA vectors (Gurunathan et al., (1997) J Exp Med 186:1137-1147; Piedrafita et al., (1999) J Immunol 163:1467-1472; and Mendez et al., (2001) J Immunol 166:5122-5128) or by administration of proteins formulated with Thl-inducing adjuvants including IL-12 (Afonso et al., (1994) Science 263:235-237; Stobie et al., (2000) Proc Natl Acad Sci USA 97:8427-8432; and Kenney et al., (1999) J Immunol 163:4481-4488) or TLR ligands such as CpG oligonucleotides (Rhee et al., (2002) J Exp Med 195:1565-1573; Stacey and Blackwell.
  • SMT Sterol 24-c-methyltransferase
  • ergosterol a target molecule of leishmanicidal and fungicidal amphotericin B (Pourshafie et al., (2004) Antimicrob Agents Chemother 48:2409-2414).
  • Amphotericin B shows selective killing activity selectively against some protozoan parasites and fungi, as ergosterol is not found in mammalian cells.
  • SMT is found in several parasites, fungi and plants, but is absent in mammals.
  • compositions of the invention employ polypeptides comprising at least an immunogenic portion of a Leishmania sterol 24-c-methyltransferase (SMT) polypeptide, or a variant thereof, wherein the portion or variant retains the desired immunogenic properties of a full length SMT polypeptide.
  • SMT Leishmania sterol 24-c-methyltransferase
  • the Leishmania SMT polypeptide comprises an amino acid sequence of an L. donovani, L infantum, L. major, or L. braziliensis SMT polypeptide.
  • the SMT polypeptide comprises an amino acid sequence set forth in any one of SEQ ID NOs: 1, 3, 5, and 11.
  • compositions comprising polynucleotides encoding the above polypeptides are provided, as well as recombinant expression vectors comprising these polynucleotide sequences and host cells transformed or transfected with such expression vectors.
  • the polynucleotide comprises a nucleic acid sequence encoding an L. donovani, L. infantum, L. major, or L. braziliensis SMT polypeptide.
  • the SMT polynucleotide comprises a nucleic acid sequence set forth in any one of SEQ ID NOs: 2, 4, 6, and 12.
  • the present invention provides a fusion polypeptide comprising at least an immunogenic portion of a Leishmania SMT antigen, and further comprising one or more heterologous fusion partners.
  • polynucleotides encoding such fusion proteins are also provided.
  • the invention provides pharmaceutical compositions which comprise one or more of the polypeptides and/or fusion polypeptides described herein, or a polynucleotide encoding such polypeptides, in combination with a physiologically acceptable carrier.
  • vaccine compositions which comprise one or more of the polypeptides and/or fusion polypeptides described herein, or a polynucleotide encoding such polypeptides, in combination with an immunostimulant.
  • the immunostimulant is an adjuvant that induces a predominately ThI type immune response.
  • the present invention provides methods for stimulating a cellular and/or humoral immune response in a patient, comprising administering to a patient a pharmaceutical composition or vaccine as described above.
  • the present invention provides methods for inducing protective immunity against leishmaniasis in a patient, comprising administering to a patient a pharmaceutical composition or vaccine as described above.
  • methods for treating a patient afflicted with leishmaniasis comprising administering to a patient a pharmaceutical composition or vaccine as described above.
  • Figure IA- IB shows the amino acid sequence alignment of SMTs from
  • L. infantum SEQ ID NO: I
  • L. donovani SEQ ID NO: 3
  • L. major SEQ ID NO: 5
  • T. cruzi SEQ ID NO: 7
  • L braziliensis SEQ ID NO: II
  • Figure 2 shows a Coomassie blue stained SDS/4-20% polyacrylamide gradient gels of uninduced E. coli Iy sates (lane 1), induced Iy sates (lane 2) and purified L. infantum SMT (lane 3). Sizes are shown in kDa on the left.
  • Figure 3 shows expression of SMT by promastigotes of L. donovani
  • Figure 4 shows the antibody responses of VL patients to SMT.
  • Figure 5 shows the antibody responses of immunized mice to SMT.
  • Levels of anti-SMT IgGl and IgG2a of mice inoculated with saline, MPL ® -SE alone, rSMT alone or rSMT plus MPL ® -SE were evaluated by ELISA. Endpoint titers were calculated by using an OD value of 0.1 as a cutoff. Three mice were used per group and means and SEM of endpoint titers of each group are shown.
  • Figure 6 shows cytokine production by immunized mice in stimulation with leishmanial antigens.
  • FIG. 7 shows the results of flow cytometric analysis of SMT-specific T-cells.
  • A A representative of flow cytometry data.
  • B TNF- ⁇ , IL-2 and IFN- ⁇ production by CD4 + and CD8 + T-cells in response to SMT. Spleen cells from mice administrated with saline, MPL-SE alone, rSMT alone and rSMT plus MPL-SE were incubated with medium alone or in a presence of rSMT, and cytokine production was analyzed by flow cytometry.
  • C Single-cell analysis of CD4 + (left) and CD8 + (right) T- cells producing multiple ThI -type cytokines.
  • Figure 8 shows the protection against L. infantum infection by SMT immunization.
  • Mice inoculated with saline, MPL ® -SE or rSMT + MPL ® -SE were challenged with L. infantum and the numbers of parasites in spleens (A) and livers (B) were measured four weeks after the infection. Mean and SEM of five mice in each group are shown. *P ⁇ 0.05 and **/ ) ⁇ 0.01 by unpaired t-test compared with both saline and MPL ® -SE groups. This is a representative of three independent experiments with similar results.
  • Figure 9 shows the protection against L. major infection by SMT immunization.
  • Mice inoculated with saline or rSMT + MPL ® -SE were challenged with L. major in the ear.
  • A Lesion development at the infection site was monitored for eight weeks. Mean and SEM of five mice in each group are shown.
  • B At eight weeks post-infection, the numbers of parasites in the infected ears were evaluated by limiting dilution assay. Bars represent the mean values of individual groups. This experiment is representative of three independent experiments with similar results.
  • SEQ ID NO: 1 is the amino acid sequence for the L. infantum sterol 24- c-methyltransferase (SMT) polypeptide.
  • SEQ ID NO: 2 is a nucleic acid sequence encoding the polypeptide of SEQ ID NO: 1.
  • SEQ ID NO: 3 is the amino acid sequence for the L. donovani sterol 24- c-methyltransferase (SMT) polypeptide.
  • SEQ ID NO: 4 is a nucleic acid sequence encoding the polypeptide of SEQ ID NO: 3.
  • SEQ ID NO: 5 is the amino acid sequence for the L. major sterol 24-c- methyltransferase (SMT) polypeptide.
  • SEQ ID NO: 6 is a nucleic acid sequence encoding the polypeptide of SEQ ID NO: 5.
  • SEQ ID NO: 7 is the amino acid sequence for the T.cruzi sterol 24-c- methyltransferase (SMT) polypeptide.
  • SEQ ID NO: 8 is the amino acid sequence for the C. albicans sterol 24- c-methyltransferase (SMT) polypeptide.
  • SEQ ID NOs: 9-10 are primers used to amplify an open reading frame from L. infantum SMT.
  • SEQ ID NO: 11 is the amino acid sequence for the L. braziliensis sterol 24-c-methyltransferase (SMT) polypeptide.
  • SEQ ID NO: 12 is a nucleic acid sequence encoding the polypeptide of
  • compositions of the invention include, for example, polypeptides that comprise at least an immunogenic portion of a Leishmania sterol 24-c-methyltransferase (SMT) polypeptide, or a variant of such a polypeptide, wherein the portion or variant retain substantially the same or similar immunogenic properties as a full length SMT polypeptide.
  • SMT Leishmania sterol 24-c-methyltransferase
  • immunization strategies using compositions of the invention provide significant in vivo protection against L. infantum, a causative agent of VL in humans and dogs.
  • the prophylactic effect achieved using compositions of the invention shows substantial improvements and advantages relative to previously reported vaccine strategies.
  • polypeptide encompasses amino acid chains of any length, including full length proteins, wherein the amino acid residues are linked by covalent bonds.
  • a polypeptide comprising an immunogenic portion of an SMT polypeptide may consist solely of an immunogenic portion, may contain two or more immunogenic portions and/or may contain additional sequences.
  • the additional sequences may be derived from a native Leishmania SMT polypeptide or may be heterologous, and such heterologous sequences may (but need not) be immunogenic.
  • An immunogenic portion of a Leishmania SMT polypeptide is a portion that is capable of eliciting an immune response (i.e., cellular and/or humoral) in a presently or previously Leishmania-m ' fected patient (such as a human or a dog) and/or in cultures of lymph node cells or peripheral blood mononuclear cells (PBMC) isolated from presently or previously Leishmania-m ' fQcted individuals.
  • the cells in which a response is elicited may comprise a mixture of cell types or may contain isolated component cells (including, but not limited to, T-cells, NK cells, macrophages, monocytes and/or B cells).
  • immunogenic portions are capable of inducing T-cell proliferation and/or a dominantly ThI -type cytokine response (e.g., IL-2, IFN- ⁇ , and/or TNF- ⁇ production by T-cells and/or NK cells; and/or IL- 12 production by monocytes, macrophages and/or B cells).
  • a dominantly ThI -type cytokine response e.g., IL-2, IFN- ⁇ , and/or TNF- ⁇ production by T-cells and/or NK cells; and/or IL- 12 production by monocytes, macrophages and/or B cells.
  • Immunogenic portions of the antigens described herein may generally be identified using techniques known to those of ordinary skill in the art, including the representative methods provided herein.
  • Immunogenic portions of an SMT polypeptide can be essentially any length provided they retain one or more of the immunogenic regions of SMT that are responsible for and/or contribute to the in vivo protection provided against leishmaniasis by the full length SMT polypeptide, as disclosed herein.
  • the ability of an immunogenic portion to react with antigen-specific antisera may be enhanced or unchanged, relative to the native protein, or may be diminished by less than 50%, and preferably less than 20%, relative to the native protein.
  • Illustrative portions will generally be at least 10, 15, 25, 50, or 100 amino acids in length, or more, up to and including full length SMT polypeptide.
  • an immunogenic portion of an SMT polypeptide is one capable of providing protection, for example in an in vivo assay as described herein, against a Leishmania species from the L. donovani complex, i.e. L. donovani and/or L. infantum, which are believed to be causative agents of VL in humans and dogs.
  • compositions and methods of the present invention also encompass variants of the above polypeptides.
  • a polypeptide "variant,” as used herein, is a polypeptide that differs from a native protein in one or more substitutions, deletions, additions and/or insertions, such that the desired immunogenicity of the variant polypeptide is not substantially diminished relative to a native SMT polypeptide.
  • the ability of a variant to react with antigen-specific antisera may be enhanced or unchanged, relative to the native protein, or may be diminished by less than 50%, and preferably less than 20%, relative to the native protein.
  • a variant of an SMT polypeptide is one capable of providing protection, for example in an in vivo assay as described herein, against a Leishmania species from the L. donovani complex, i.e. L. donovani and/or L. infantum.
  • Variants may generally be identified by modifying one of the above polypeptide sequences and evaluating the reactivity of the modified polypeptide with antigen-specific antibodies or antisera as described herein or by performing an in vivo protection assay as described herein.
  • Polypeptide variants will generally include those exhibiting at least about 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identity (for example, determined as described below) to an SMT polypeptide disclosed herein. In certain embodiments, a variant will contain conservative substitutions.
  • a “conservative substitution” is one in which an amino acid is substituted for another amino acid that has similar properties, such that one skilled in the art of peptide chemistry would expect the secondary structure and hydropathic nature of the polypeptide to be substantially unchanged.
  • Amino acid substitutions may generally be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity and/or the amphipathic nature of the residues.
  • negatively charged amino acids include aspartic acid and glutamic acid; positively charged amino acids include lysine and arginine; and amino acids with uncharged polar head groups having similar hydrophilicity values include leucine, isoleucine and valine; glycine and alanine; asparagine and glutamine; and serine, threonine, phenylalanine and tyrosine.
  • variant polypeptides differ from a native sequence by substitution, deletion or addition of five amino acids or fewer.
  • Variants may also (or alternatively) be modified by, for example, the deletion or addition of amino acids that have minimal influence on the immunogenicity, secondary structure and hydropathic nature of the polypeptide.
  • Polynucleotides may comprise a native sequence (i.e., an endogenous sequence that encodes a protein or a portion thereof) or may comprise a variant, or a biological or antigenic functional equivalent of such a sequence.
  • Polynucleotide variants may contain one or more substitutions, additions, deletions and/or insertions, as further described below, preferably such that the immunogenicity of the encoded polypeptide is not substantially diminished, relative to a native tumor protein. The effect on the immunogenicity of the encoded polypeptide may generally be assessed as described herein.
  • two sequences are said to be “identical” if the sequence of nucleotides or amino acids in the two sequences is the same when aligned for maximum correspondence, as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity.
  • a “comparison window” as used herein refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • Alignment of sequences for comparison may be conducted using, for example, the Megalign program in the Lasergene suite of bioinformatics software (DNASTAR, Inc., Madison, WI) using default parameters.
  • This program embodies several alignment schemes described in the following references: Dayhoff, M. O. (1978) A model of evolutionary change in proteins - Matrices for detecting distant relationships. In Dayhoff, M. O. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington DC Vol. 5, Suppl. 3, pp. 345-358; Hein (1990) Unified Approach to Alignment and Phylogenes pp. 626-645 Methods in Enzymology vol.
  • alignment of sequences for comparison may be conducted by the local identity algorithm of Smith and Waterman (1981) Add. APL Math 2:482, by the identity alignment algorithm of Needleman and Wunsch (1970) J MoI. Biol.
  • GCG Genetics Computer Group
  • BLAST and BLAST 2.0 are described in Altschul et al. (1977) Nucl. Acids Res. 25:3389-3402 and Altschul et al. (1990) J. MoI. Biol. 215:403-410, respectively.
  • BLAST and BLAST 2.0 can be used, for example with the parameters described herein, to determine percent sequence identity for the polynucleotides and polypeptides of the invention.
  • Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information.
  • cumulative scores can be calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always ⁇ 0).
  • M forward score for a pair of matching residues; always >0
  • N penalty score for mismatching residues; always ⁇ 0
  • a scoring matrix can be used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached.
  • the BLAST algorithm parameters W, T and X determine the sensitivity and speed of the alignment.
  • the "percentage of sequence identity” is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide or polypeptide sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • additions or deletions i.e., gaps
  • the percentage is calculated by determining the number of positions at which the identical nucleic acid bases or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e., the window size) and multiplying the results by 100 to yield the percentage of sequence identity.
  • the present invention encompasses polynucleotide and polypeptide sequences having substantial identity to the sequences disclosed herein, for example those comprising at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% or higher, sequence identity compared to a polynucleotide or polypeptide sequence of this invention using the methods described herein, (e.g., BLAST analysis using standard parameters, as described below).
  • BLAST analysis using standard parameters, as described below.
  • the present invention provides isolated polynucleotides and polypeptides comprising various lengths of contiguous stretches of sequence identical to or complementary to one or more of the sequences disclosed herein.
  • polynucleotides are provided by this invention that comprise at least about 15, 20, 30, 40, 50, 75, 100, 150, 200, 300, 400, 500 or 1000 or more contiguous nucleotides of one or more of the sequences disclosed herein as well as all intermediate lengths there between.
  • intermediate lengths in this context, means any length between the quoted values, such as 16, 17, 18, 19, etc.; 21, 22, 23, etc.; 30, 31, 32, etc.
  • polynucleotides of the present invention, or fragments thereof, regardless of the length of the coding sequence itself, may be combined with other DNA sequences, such as promoters, polyadenylation signals, additional restriction enzyme sites, multiple cloning sites, other coding segments, and the like, such that their overall length may vary considerably. It is therefore contemplated that a nucleic acid fragment of almost any length may be employed, with the total length preferably being limited by the ease of preparation and use in the intended recombinant DNA protocol.
  • illustrative DNA segments with total lengths of about 10,000, about 5000, about 3000, about 2,000, about 1,000, about 500, about 200, about 100, about 50 base pairs in length, and the like, (including all intermediate lengths) are contemplated to be useful in many implementations of this invention.
  • the present invention is directed to polynucleotides that are capable of hybridizing under moderately stringent conditions to a polynucleotide sequence provided herein, or a fragment thereof, or a complementary sequence thereof.
  • Hybridization techniques are well known in the art of molecular biology.
  • suitable moderately stringent conditions for testing the hybridization of a polynucleotide of this invention with other polynucleotides include prewashing in a solution of 5 X SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0); hybridizing at 50°C-65°C, 5 X SSC, overnight; followed by washing twice at 65 0 C for 20 minutes with each of 2X, 0.5X and 0.2X SSC containing 0.1% SDS.
  • nucleotide sequences that encode a polypeptide as described herein. Some of these polynucleotides bear minimal homology to the nucleotide sequence of any native gene. Nonetheless, polynucleotides that vary due to differences in codon usage are specifically contemplated by the present invention. Further, alleles of the genes comprising the polynucleotide sequences provided herein are within the scope of the present invention. Alleles are endogenous genes that are altered as a result of one or more mutations, such as deletions, additions and/or substitutions of nucleotides. The resulting mRNA and protein may, but need not, have an altered structure or function. Alleles may be identified using standard techniques (such as hybridization, amplification and/or database sequence comparison).
  • fusion polypeptides comprise at least an immunogenic portion of an SMT polypeptide and further comprising a heterologous fusion partner, as well as polynucleotides encoding such fusion polypeptides.
  • a fusion polypeptide comprises one or more immunogenic portions of an SMT polypeptide and one or more additional immunogenic Leishmania sequences, which are joined via a peptide linkage into a single amino acid chain.
  • a fusion polypeptide may comprise multiple Leishmania antigenic epitopes wherein at least one of the epitopes is from an SMT polypeptide.
  • an epitope is a portion of an antigen that reacts with blood samples from Leishmania-infccted individuals (i.e., an epitope is specifically bound by one or more antibodies and/or T-cells present in such blood samples.
  • a heterologous fusion partner comprises a sequence that assists in providing T helper epitopes (an immunological fusion partner), preferably T helper epitopes recognized by humans, or that assists in expressing the protein (an expression enhancer) at higher yields than the native recombinant protein.
  • an immunological fusion partner preferably T helper epitopes recognized by humans, or that assists in expressing the protein (an expression enhancer) at higher yields than the native recombinant protein.
  • Certain preferred fusion partners include both immunological and expression-enhancing fusion partners.
  • Other fusion partners may be selected so as to increase the solubility of the protein or to enable the protein to be targeted to desired intracellular compartments.
  • Still further fusion partners include affinity tags, which facilitate purification of the protein.
  • an immunological fusion partner comprises a sequence derived from protein D, a surface protein of the gram-negative bacterium Haemophilus influenza B (WO 91/18926).
  • one protein D derivative comprises approximately the first third of the protein (e.g., the first N- terminal 100 110 amino acids), and a protein D derivative may be lipidated.
  • the first 109 residues of a lipoprotein D fusion partner is included on the N-terminus to provide the polypeptide with additional exogenous T cell epitopes and to increase the expression level in E. coli (thus functioning as an expression enhancer).
  • the lipid tail ensures optimal presentation of the antigen to antigen presenting cells.
  • illustrative fusion partners include the non-structural protein from influenzae virus, NSl (hemaglutinin). Typically, the N-terminal 81 amino acids are used, although different fragments that include T-helper epitopes may also be used.
  • an immunological fusion partner comprises an amino acid sequence derived from the protein known as LYTA, or a portion thereof (preferably a C-terminal portion).
  • LYTA is derived from Streptococcus pneumoniae, which synthesizes an N-acetyl-L-alanine amidase known as amidase LYTA (encoded by the LytA gene; Gene (1986) 43:265-292).
  • LYTA is an autolysin that specifically degrades certain bonds in the peptidoglycan backbone.
  • the C-terminal domain of the LYTA protein is responsible for the affinity to the choline or to some choline analogues such as DEAE. This property has been exploited for the development of E.
  • coli C-LYTA expressing plasmids useful for expression of fusion proteins. Purification of hybrid proteins containing the C-LYTA fragment at the amino terminus has been described (see Biotechnology (1992) 10:795-798). Within a particular embodiment, a repeat portion of LYTA may be incorporated into a fusion protein. A repeat portion is found in the C-terminal region starting at residue 178. A more particular repeat portion incorporates residues 188-305.
  • Fusion sequences may be joined directly (i.e., with no intervening amino acids) or may be joined by way of a linker sequence (e.g., Gly-Cys-Gly) that does not significantly diminish the immunogenic properties of the component polypeptides.
  • the polypeptides forming the fusion protein are typically linked C-terminus to N-terminus, although they can also be linked C-terminus to C-terminus, N-terminus to N-terminus, or N-terminus to C-terminus.
  • the polypeptides of the fusion protein can be in any order.
  • Fusion polypeptides or fusion proteins can also include conservatively modified variants, polymorphic variants, alleles, mutants, subsequences, interspecies homologs, and immunogenic fragments of the antigens that make up the fusion protein.
  • Fusion polypeptides may generally be prepared using standard techniques, including recombinant technology, chemical conjugation, and the like. For example, DNA sequences encoding the polypeptide components of a fusion may be assembled separately and ligated into an appropriate expression vector. The 3' end of the DNA sequence encoding one polypeptide component is ligated, with or without a peptide linker, to the 5' end of a DNA sequence encoding the second polypeptide component so that the reading frames of the sequences are in frame. This permits translation into a single fusion polypeptide that retains the biological activity of the component polypeptides.
  • a peptide linker sequence may be employed to separate the fusion components by a distance sufficient to ensure that each polypeptide folds into its desired secondary and/or tertiary structures.
  • Such a peptide linker sequence may be incorporated into the fusion polypeptide using standard techniques well known in the art.
  • Suitable peptide linker sequences may be chosen, for example, based on one or more of the following factors: (1) their ability to adopt a flexible extended conformation; (2) their inability to adopt a secondary structure that could interact with functional epitopes on the first and second polypeptides; and (3) the lack of hydrophobic or charged residues that might react with the polypeptide functional epitopes.
  • Certain preferred peptide linker sequences contain GIy, Asn and Ser residues.
  • linker sequences which may be usefully employed as linkers include those disclosed in Maratea et al., (1985) Gene 40:39-46,; Murphy et al., (1986) Proc. Natl. Acad. ScL USA 83:8258-8262; U.S. Patent No. 4,935,233 and U.S. Patent No. 4,751,180.
  • the linker sequence may generally be from 1 to about 50 amino acids in length. Linker sequences are not required when the first and second polypeptides have non-essential N-terminal amino acid regions that can be used to separate the functional domains and prevent steric interference.
  • the ligated DNA sequences are operably linked to suitable transcriptional or translational regulatory elements.
  • the regulatory elements responsible for expression of DNA are located only 5' to the DNA sequence encoding the first polypeptides.
  • stop codons required to end translation and transcription termination signals are only present 3' to the DNA sequence encoding the second polypeptide.
  • polypeptides and fusion polypeptides are isolated.
  • An "isolated" polypeptide or polynucleotide is one that is removed from its original environment.
  • a naturally-occurring protein is isolated if it is separated from some or all of the coexisting materials in the natural system.
  • polypeptides are at least about 90% pure, more preferably at least about 95% pure and most preferably at least about 99% pure.
  • a polynucleotide is considered to be isolated if, for example, it is cloned into a vector that is not a part of the natural environment.
  • Leishmania SMT polypeptides and polynucleotides of the invention may be prepared or isolated using any of a variety of procedures and using any of a variety of Leishmania species including, but not limited to, L donovani, L. chagasi, L. infantum, L. major, L. amazonensis, L. braziliensis, L panamensis, L mexicana, L. tropica, and L guyanensis. Such species are available, for example, from the American Type Culture Collection (ATCC), Rockville, MD.
  • ATCC American Type Culture Collection
  • the SMT polypeptides described herein are preferably immunogenic.
  • the polypeptides (and immunogenic portions thereof) are capable of eliciting an immune response in cultures of lymph node cells and/or peripheral blood mononuclear cells (PBMC) isolated from presently or previously Leishmania-infected individuals.
  • PBMC peripheral blood mononuclear cells
  • the antigens, and immunogenic portions thereof have the ability to induce T-cell proliferation and/or to elicit a dominantly ThI -type cytokine response (e.g., IL-2, IFN- ⁇ , and/or TNF-oc production by T-cells and/or NK cells; and/or IL- 12 production by monocytes, macrophages and/or B cells) in cells isolated from presently or previously Leishmania- infected individuals.
  • ThI -type cytokine response e.g., IL-2, IFN- ⁇ , and/or TNF-oc production by T-cells and/or NK cells; and/or IL- 12 production by monocytes, macrophages and/or B cells
  • a Leishmania-m ' fQCted individual may be afflicted with a form of leishmaniasis (such as subclinical, cutaneous, mucosal or active visceral) or may be asymptomatic.
  • Such individuals may be identified using methods known to those of ordinary skill in the art.
  • Individuals with leishmaniasis may be identified based on clinical findings associated with at least one of the following: isolation of parasite from lesions, a positive skin test with Leishmania lysate or a positive serological test.
  • Asymptomatic individuals are infected individuals who have no signs or symptoms of the disease.
  • Such individuals can be identified based on a positive serological test and/or skin test with Leishmania lysate.
  • PBMC which refers to a preparation of nucleated cells consisting primarily of lymphocytes and monocytes that are present in peripheral blood, encompasses both mixtures of cells and preparations of one or more purified cell types.
  • PBMC may be isolated by methods known to those in the art.
  • PBMC may be isolated by density centrifugation through, for example, Ficoll TM (Winthrop Laboratories, New York).
  • Lymph node cultures may generally be prepared by immunizing BALB/c mice (e.g., in the rear foot pad) with Leishmania promastigotes emulsified in complete Fre ⁇ nd's adjuvant.
  • the draining lymph nodes may be excised following immunization and T-cells may be purified in an anti-mouse Ig column to remove the B cells, followed by a passage through a Sephadex GlO column to remove the macrophages.
  • lymph node cells may be isolated from a human following biopsy or surgical removal of a lymph node.
  • the ability of a Leishmania SMT polypeptide, or a portion, variant or fusion thereof, to induce a response in PBMC or lymph node cell cultures may be evaluated, for example, by contacting the cells with the polypeptide and measuring a suitable response.
  • the amount of polypeptide that is sufficient for the evaluation of about 2 x 10 5 cells ranges from about 10 ng to about 100 ⁇ g, and preferably is about 1-10 ⁇ g.
  • the incubation of polypeptide with cells is typically performed at 37°C for about 1-3 days. Following incubation with polypeptide, the cells are assayed for an appropriate response. If the response is a proliferative response, any of a variety of techniques well known to those of ordinary skill in the art may be employed.
  • the cells may be exposed to a pulse of radioactive thymidine and the incorporation of label into cellular DNA measured.
  • a polypeptide that results in at least a three fold increase in proliferation above background i.e., the proliferation observed for cells cultured without polypeptide is considered to be able to induce proliferation.
  • the response to be measured may be the secretion of one or more cytokines (such as interferon- ⁇ (IFN- ⁇ ), interleukin-4 (IL-4), interleukin-12 (p70 and/or p40), interleukin-2 (IL-2) and/or tumor necrosis factor- ⁇ (TNF- ⁇ )) or the change in the level of mRNA encoding one or more specific cytokines.
  • cytokines such as interferon- ⁇ (IFN- ⁇ ), interleukin-4 (IL-4), interleukin-12 (p70 and/or p40), interleukin-2 (IL-2) and/or tumor necrosis factor- ⁇ (TNF- ⁇ )
  • the secretion of interferon- ⁇ , interleukin-2, tumor necrosis factor- ⁇ and/or interleukin-12 is indicative of a ThI response, which is responsible for the protective effect against Leishmania.
  • Assays for any of the above cytokines may generally be performed using methods known to those of ordinary skill in the art, such as an enzyme-linked immunosorbent assay (ELISA).
  • ELISA enzyme-linked immunosorbent assay
  • Suitable antibodies for use in such assays may be obtained from a variety of sources such as Chemicon, Temucula, CA and PharMingen, San Diego, CA, and may generally be used according to the manufacturer's instructions.
  • the level of mRNA encoding one or more specific cytokines may be evaluated by, for example, amplification by polymerase chain reaction (PCR).
  • a polypeptide that is able to induce, in a preparation of about 1-3 x 10 5 cells, the production of 30 pg/mL of IL-12, IL-4, IFN- ⁇ , TNF- ⁇ or IL-12 p40, or 10 pg/mL of IL-12 p70, is considered able to stimulate production of a cytokine.
  • Immunogenic portions of a Leishmania SMT polypeptide may be prepared and evaluated using well known techniques, such as those summarized in Paul, Fundamental Immunology, 3rd ed., 243-247 (Raven Press, 1993) and references cited therein. Such techniques include screening polypeptides derived from the native antigen for immunogenic properties using, for example, a representative techniques described herein.
  • SMT polypeptides, portions and variants may be generated by synthetic or recombinant means.
  • Synthetic polypeptides having fewer than about 100 amino acids, and generally fewer than about 50 amino acids may be generated using techniques well known to those of ordinary skill in the art.
  • such polypeptides may be synthesized using any of the commercially available solid-phase techniques, such as the Merrifield solid-phase synthesis method, where amino acids are sequentially added to a growing amino acid chain. See Merrifield, (1963) J Am. Chem. Soc. 85:2149-2146.
  • Equipment for automated synthesis of polypeptides is commercially available from suppliers such as Perkin Elmer/Applied BioSystems Division, Foster City, CA, and may be operated according to the manufacturer's instructions.
  • Recombinant polypeptides containing portions and/or variants of a native SMT polypeptide may be readily prepared from a DNA sequence encoding the antigen, using well known and established techniques. For example, superaatants from suitable host/vector systems which secrete recombinant protein into culture media may be first concentrated using a commercially available filter. Following concentration, the concentrate may be applied to a suitable purification matrix such as an affinity matrix or an ion exchange resin. In general, any of a variety of expression vectors known to those of ordinary skill in the art may be employed to express recombinant polypeptides of this invention.
  • Expression may be achieved in any appropriate host cell that has been transformed or transfected with an expression vector containing a polynucleotide that encodes a recombinant polypeptide.
  • Suitable host cells include prokaryotes, yeast and higher eukaryotic cells.
  • the host cells employed are E. coli, yeast or a mammalian cell line such as COS or CHO.
  • the DNA sequences expressed in this manner may encode naturally occurring antigens, portions of naturally occurring antigens, or other variants thereof.
  • variants of a native antigen may generally be prepared using standard mutagenesis techniques, such as oligonucleotide- directed site-specific mutagenesis, and sections of the DNA sequence may be removed to permit preparation of truncated polypeptides.
  • polypeptides, polynucleotides, portions, variants, fusion polypeptides, etc., as described herein are incorporated into pharmaceutical compositions or vaccines.
  • Pharmaceutical compositions generally comprise one or more polypeptides, polynucleotides, portions, variants, fusion polypeptides, etc., as described herein, in combination with a physiologically acceptable carrier.
  • Vaccines also referred to as immunogenic compositions, generally comprise one or more of the polypeptides, polynucleotides, portions, variants, fusion proteins, etc., as described herein, in combination with an immunostimulant, such as an adjuvant.
  • An immunostimulant may be any substance that enhances or potentiates an immune response (antibody and/or cell-mediated) to an exogenous antigen.
  • immunostimulants include adjuvants, biodegradable microspheres (e.g., polylactic galactide) and liposomes (into which the compound is incorporated; see, e.g., Fullerton, U.S. Pat. No. 4,235,877).
  • Vaccine preparation is generally described in, for example, Powell & Newman, eds., Vaccine Design (the subunit and adjuvant approach) (1995).
  • immunostimulants may be employed in the vaccines of this invention.
  • an adjuvant may be included.
  • Many adjuvants contain a substance designed to protect the antigen from rapid catabolism, such as aluminum hydroxide or mineral oil, and a stimulator of immune responses, such as lipid A (natural or synthetic), Bortadella pertussis or Mycobacterium species or Mycobacterium-de ⁇ ved proteins.
  • Suitable adjuvants are commercially available as, for example, Freund's Incomplete Adjuvant and Complete Adjuvant (Difco Laboratories, Detroit, Mich.); Merck Adjuvant 65 (Merck and Company, Inc., Rahway, N.J.); AS-2 and derivatives thereof (GlaxoSmithKline Beecham, Philadelphia, Pa.); CWS, TDM, Leif, aluminum salts such as aluminum hydroxide gel (alum) or aluminum phosphate; salts of calcium, iron or zinc; an insoluble suspension of acylated tyrosine; acylated sugars; cationically or anionically derivatized polysaccharides; polyphosphazenes; biodegradable microspheres; monophosphoryl lipid A and quil A. Cytokines, such as GM-CSF or interleukin-2, -7, or -12, may also be used as adjuvants. Other illustrative adjuvants useful in the context of the invention include
  • Toll-like receptor agonists such as TLR7 agonists, TLR7/8 agonists, and the like.
  • Still other illustrative adjuvants include imiquimod, gardiquimod, resiquimod, and related compounds.
  • Certain preferred vaccines employ adjuvant systems designed to induce an immune response predominantly of the ThI type.
  • Th2-type cytokines e.g., IL-4, IL-5, IL-6 and IL-IO
  • a patient will support an immune response that includes ThI- and Th2-type responses.
  • Th2-type responses in which a response is predominantly Thl-type, the level of
  • Thl-type cytokines will increase to a greater extent than the level of Th2-type cytokines.
  • the levels of these cytokines may be readily assessed using standard assays.
  • Certain adjuvants for use in eliciting a predominantly Thl-type response include, for example, a combination of monophosphoryl lipid A, preferably 3-de-O- acylated monophosphoryl lipid A (3D-MPLTM), together with an aluminum salt (U.S. Pat. Nos. 4,436,727; 4,877,611 ; 4,866,034; and 4,912,094).
  • CpG-containing oligonucleotides in which the CpG dinucleotide is unmethylated also induce a predominantly ThI response.
  • Such oligonucleotides are well known and are described, for example, in WO 96/02555, WO 99/33488 and U.S. Pat. Nos.
  • Another illustrative adjuvant comprises a saponin, such as Quil A, or derivatives thereof, including QS21 and QS7 (Aquila Biopharmaceuticals Inc., Framingham, Mass.); Escin; Digitonin; or Gypsophila or Chenopodium quinoa saponins.
  • Other illustrative formulations include more than one saponin in the adjuvant combinations of the present invention, for example combinations of at least two of the following group comprising QS21, QS7, Quil A, ⁇ - escin, or digitonin.
  • the adjuvant system includes the combination of a monophosphoryl lipid A and a saponin derivative, such as the combination of QS21 and 3D-MPLTM adjuvant, as described in WO 94/00153, or a less reactogenic composition where the QS21 is quenched with cholesterol, as described in WO 96/33739.
  • Other formulations comprise an oil-in-water emulsion and tocopherol.
  • Another adjuvant formulation employing QS21, 3D-MPLTM adjuvant and tocopherol in an oil-in-water emulsion is described in WO 95/17210.
  • Another enhanced adjuvant system involves the combination of a CpG- containing oligonucleotide and a saponin derivative as disclosed in WO 00/09159.
  • the adjuvant used in the present invention is a glucopyranosyl lipid A (GLA) adjuvant, as described in pending U.S. Patent Application Publication No. 20080131466, the disclosure of which is incorporated herein by reference in its entirety.
  • GLA glucopyranosyl lipid A
  • illustrative adjuvants include Montanide ISA 720 (Seppic, France), SAF (Chiron, Calif, United States), ISCOMS (CSL), MF-59 (Chiron), the SBAS series of adjuvants (e.g., SBAS-2, AS2', AS2," SBAS-4, or SBAS6, available from SmithKline Beecham, Rixensart, Belgium), Detox, RC-529 (Corixa, Hamilton, Mont.) and other aminoalkyl glucosaminide 4-phosphates (AGPs), such as those described in pending U.S. patent application Ser. Nos.
  • compositions of the invention may also, or alternatively, comprise T cells specific for a Leishmania SMT antigen.
  • T cells may generally be prepared in vitro or ex vivo, using standard procedures.
  • T cells may be isolated from bone marrow, peripheral blood, or a fraction of bone marrow or peripheral blood of a patient.
  • T cells may be derived from related or unrelated humans, non- human mammals, cell lines or cultures.
  • T cells may be stimulated with a polypeptide of the invention, polynucleotide encoding such a polypeptide, and/or an antigen presenting cell (APC) that expresses such a polypeptide.
  • APC antigen presenting cell
  • Such stimulation is performed under conditions and for a time sufficient to permit the generation of T cells that are specific for the polypeptide.
  • the polypeptide or polynucleotide is present within a delivery vehicle, such as a microsphere, to facilitate the generation of specific T cells.
  • T cells are considered to be specific for a polypeptide of the invention if the T cells specifically proliferate, secrete cytokines or kill target cells coated with the polypeptide or expressing a gene encoding the polypeptide.
  • T cell specificity may be evaluated using any of a variety of standard techniques. For example, within a chromium release assay or proliferation assay, a stimulation index of more than two fold increase in lysis and/or proliferation, compared to negative controls, indicates T cell specificity. Such assays may be performed, for example, as described in Chen et al., (1994) Cancer Res. 54:1065-1070). Alternatively, detection of the proliferation of
  • T cells may be accomplished by a variety of known techniques.
  • T cell proliferation can be detected by measuring an increased rate of DNA synthesis (e.g., by pulse-labeling cultures of T cells with tritiated thymidine and measuring the amount of tritiated thymidine incorporated into DNA).
  • a polypeptide of the invention lOOng/ml-lOO ⁇ g/ml, preferably 200ng/ml-25 ⁇ g/ml
  • contact as described above for 2-3 hours should result in activation of the T cells, as measured using standard cytokine assays in which a two fold increase in the level of cytokine release
  • TNF TNF or IFN- ⁇
  • TNF TNF or IFN- ⁇
  • TNF or IFN- ⁇ TNF or IFN- ⁇
  • TNF or IFN- ⁇ TNF or IFN- ⁇
  • TNF or IFN- ⁇ TNF or IFN- ⁇
  • T cells that have been activated in response to a polypeptide, polynucleotide or polypeptide-expressing APC may be CD4+ and/or CD8+.
  • Protein-specific T cells may be expanded using standard techniques.
  • the T cells are derived from a patient, a related donor or an unrelated donor, and are administered to the patient following stimulation and expansion.
  • compositions of the invention formulation of pharmaceutically- acceptable excipients and carrier solutions is well-known to those of skill in the art, as is the development of suitable dosing and treatment regimens for using the particular compositions described herein in a variety of treatment regimens, including e.g., oral, parenteral, intravenous, intranasal, and intramuscular administration and formulation.
  • the compositions disclosed herein may be delivered via oral administration to a subject.
  • these compositions may be formulated with an inert diluent or with an assimilable edible carrier, or they may be enclosed in hard- or soft-shell gelatin capsule, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet.
  • Solutions of the active compounds as free base or pharmacologically acceptable salts may be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose.
  • Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils.
  • compositions suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (U.S. Pat. No. 5,466,468, specifically incorporated herein by reference in its entirety).
  • sterile aqueous solutions or dispersions include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (U.S. Pat. No. 5,466,468, specifically incorporated herein by reference in its entirety).
  • the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils.
  • polyol e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • suitable mixtures thereof e.g., vegetable oils
  • vegetable oils e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • suitable mixtures thereof e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • vegetable oils e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • aqueous solution for parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
  • a sterile aqueous medium that can be employed will be known to those of skill in the art in light of the present disclosure.
  • one dosage may be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion (see, e.g., Remington's Pharmaceutical Sciences, 15th Edition, pp.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with the various other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • compositions disclosed herein may be formulated in a neutral or salt form.
  • Pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • solutions Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • the formulations are easily administered in a variety of dosage forms such as injectable solutions, drug-release capsules, and the like.
  • carrier includes any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like.
  • carrier includes any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like.
  • the use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
  • compositions that do not produce an allergic or similar untoward reaction when administered to a human.
  • pharmaceutically-acceptable refers to molecular entities and compositions that do not produce an allergic or similar untoward reaction when administered to a human.
  • aqueous composition that contains a protein as an active ingredient is well understood in the art.
  • injectables either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid prior to injection can also be prepared.
  • the preparation can also be emulsified.
  • the compositions may be delivered by intranasal sprays, inhalation, and/or other aerosol delivery vehicles.
  • Methods for delivering genes, polynucleotides, and peptide compositions directly to the lungs via nasal aerosol sprays has been described e.g., in U.S. Pat. No. 5,756,353 and U.S. Pat. No. 5,804,212 (each specifically incorporated herein by reference in its entirety).
  • the delivery of drugs using intranasal microparticle resins Takenaga et al., 1998) and lysophosphatidyl-glycerol compounds (U.S. Pat. No.
  • the delivery may occur by use of liposomes, nanocapsules, microparticles, microspheres, lipid particles, vesicles, and the like, for the introduction of the compositions of the present invention into suitable host cells.
  • the compositions of the present invention may be formulated for delivery either encapsulated in a lipid particle, a liposome, a vesicle, a nanosphere, a nanoparticle or the like.
  • the formulation and use of such delivery vehicles can be carried out using known and conventional techniques.
  • a pharmaceutical or immunogenic composition may, alternatively, contain an immunostimulant and a DNA molecule encoding one or more of the polypeptides or fusion proteins described above, such that a desired polypeptide is generated in situ.
  • the DNA may be present within any of a variety of delivery systems known to those of ordinary skill in the art, including nucleic acid expression systems, bacteria and viral expression systems. Appropriate nucleic acid expression systems contain the necessary DNA sequences for expression in the patient (such as a suitable promoter and terminating signal).
  • Bacterial delivery systems involve the administration of a bacterium (such as Bacillus-Calmette-Guerrin) that expresses an immunogenic portion of the polypeptide on its cell surface.
  • the DNA may be introduced using a viral expression system (e.g., vaccinia or other pox virus, retrovirus, or adenovirus), which may involve the use of a nonpathogenic (defective), replication competent virus.
  • a viral expression system e.g., vaccinia or other pox virus, retrovirus, or adenovirus
  • a nonpathogenic (defective), replication competent virus e.g., vaccinia or other pox virus, retrovirus, or adenovirus
  • Techniques for incorporating DNA into such expression systems are well known to those of ordinary skill in the art.
  • the DNA may also be "naked,” as described, for example, in Ulmer et al., (1993) Science 259:1745-1749 and reviewed by Cohen, (1993) Science 259:1691-1692.
  • the uptake of naked DNA may be increased by coating the DNA onto biodegradable beads, which are efficiently transported into the cells.
  • compositions and vaccines of the invention may be used, for example, to induce protective immunity against Leishmania in a patient, such as a human or a dog, to prevent leishmaniasis or diminish its severity.
  • the compositions and vaccines may also be used to stimulate an immune response, which may be cellular and/or humoral, in a patient, for treating an individual already infected.
  • the immune responses generated include a preferential ThI immune response (i.e., a response characterized by the production of the cytokines interleukin-1, interleukin-2, interleukin-12 and/or interferon- ⁇ , as well as tumor necrosis factor- ⁇ ).
  • the immune response involves production of interleukin-12 and/or interleukin- 2, or the stimulation of gamma delta T-cells.
  • the response stimulated may include IL- 12 production.
  • Such responses may also be elicited in biological samples of PBMC or components thereof derived from Leishmania-inf ected or uninfected individuals.
  • assays for any of the above cytokines, as well as other known cytokines may generally be performed using methods known to those of ordinary skill in the art, such as an enzyme-linked immunosorbent assay (ELISA).
  • ELISA enzyme-linked immunosorbent assay
  • a suitable dose is an amount of polypeptide or DNA that, when administered as described above, is capable of eliciting an immune response in an immunized patient sufficient to protect the patient from leishmaniasis for at least 1-2 years.
  • the amount of polypeptide present in a dose ranges from about 100 ng to about lmg per kg of host, typically from about 10 ⁇ g to about 100 ⁇ g.
  • Suitable dose sizes will vary with the size of the patient, but will typically range from about 0.1 mL to about 5 mL.
  • compositions, fusion polypeptides and polynucleotides are also useful as diagnostic reagents for detecting and/or monitoring Leishmania infection in a patient.
  • the compositions, fusion polypeptides, and polynucleotides of the invention may be used in in vitro and in vivo assays for detecting humoral antibodies or cell-mediated immunity against Leishmania for diagnosis of infection, monitoring of disease progression or test-of-cure evaluation.
  • the diagnostic methods and kits preferably employ an SMT polypeptide, portion, variant or the like, optionally in combination with one or more other
  • Leishmania antigens In certain embodiments, it will be preferred to use a multiple antigens as described herein, e.g., three or more, four or more, five or more, six or more, etc., in a diagnostic method of the invention.
  • the antigens may be used in essentially any assay format desired, e.g., as individual antigens assayed separately, as multiple antigens assayed simultaneously, as antigens immobilized on a solid support such as an array, or the like.
  • diagnostic kits for detecting Leishmania infection in a biological sample comprising (a) a polypeptide comprising at least an immunogenic portion of an SMT polypeptide described herein, and (b) a detection reagent.
  • Leishmania infection in a biological sample comprising (a) an antibody or antigen binding fragment thereof that is specific for a polypeptide comprising at least an immunogenic portion of an SMT polypeptide described herein, and (b) a detection reagent.
  • methods for detecting the presence of Leishmania infection in a biological sample, comprising (a) contacting a biological sample with a monoclonal antibody that binds to an SMT polypeptide described herein; and (b) detecting in the biological sample the presence of Leishmania proteins that bind to the monoclonal antibody.
  • the assay involves the use of polypeptide immobilized on a solid support to bind to and remove the antibody from the sample. The bound antibody may then be detected using a detection reagent that binds to the antibody/peptide complex and contains a detectable reporter group. Suitable detection reagents include antibodies that bind to the antibody/polypeptide complex and free polypeptide labeled with a reporter group (e.g., in a semi-competitive assay).
  • a competitive assay may be utilized, in which an antibody that binds to the polypeptide is labeled with a reporter group and allowed to bind to the immobilized antigen after incubation of the antigen with the sample.
  • the extent to which components of the sample inhibit the binding of the labeled antibody to the polypeptide is indicative of the reactivity of the sample with the immobilized polypeptide.
  • the solid support may be any solid material known to those of ordinary skill in the art to which the antigen may be attached.
  • the solid support may be a test well in a microtiter plate or a nitrocellulose or other suitable membrane.
  • the support may be a bead or disc, such as glass, fiberglass, latex or a plastic material such as polystyrene or polyvinylchloride.
  • the support may also be a magnetic particle or a fiber optic sensor, such as those disclosed, for example, in U.S. Patent No. 5,359,681.
  • the polypeptide may be bound to the solid support using any of a variety of techniques known and available in the art.
  • bound refers to both noncovalent association, such as adsorption, and covalent attachment (which may be a direct linkage between the antigen and functional groups on the support or may be a linkage by way of a cross-linking agent). Binding by adsorption to a well in a microtiter plate or to a membrane is preferred. In such cases, adsorption may be achieved by contacting the polypeptide, in a suitable buffer, with the solid support for a suitable amount of time.
  • the diagnostic assay employed is an enzyme linked immunosorbent assay (ELISA).
  • ELISA enzyme linked immunosorbent assay
  • This assay may be performed by first contacting a polypeptide antigen that has been immobilized on a solid support, commonly the well of a microtiter plate, with the sample, such that antibodies to the polypeptide within the sample are allowed to bind to the immobilized polypeptide. Unbound sample is then removed from the immobilized polypeptide and a detection reagent capable of binding to the immobilized antibody-polypeptide complex is added. The amount of detection reagent that remains bound to the solid support is then determined using a method appropriate for the specific detection reagent. Once the polypeptide is immobilized on the support, the remaining protein binding sites on the support are typically blocked.
  • ELISA enzyme linked immunosorbent assay
  • an appropriate contact time i.e., incubation time
  • incubation time is that period of time that is sufficient to detect the presence of antibody to Leishmania within an infected sample.
  • the contact time is sufficient to achieve a level of binding that is at least 95% of that achieved at equilibrium between bound and unbound antibody.
  • Unbound sample may then be removed by washing the solid support with an appropriate buffer, such as PBS containing 0.1% Tween 20TM.
  • Detection reagent may then be added to the solid support.
  • An appropriate detection reagent is any compound that binds to the immobilized antibody-polypeptide complex and that can be detected by any of a variety of means known to those in the art.
  • the detection reagent generally contains a binding agent (such as, for example, Protein A, Protein G, immunoglobulin, lectin or free antigen) conjugated to a reporter group.
  • a binding agent such as, for example, Protein A, Protein G, immunoglobulin, lectin or free antigen conjugated to a reporter group.
  • Illustrative reporter groups include enzymes (such as horseradish peroxidase), substrates, cofactors, inhibitors, dyes, radionuclides, luminescent groups, fluorescent groups and biotin.
  • binding agent to reporter group may be achieved using standard methods known to those of ordinary skill in the art.
  • the detection reagent is then incubated with the immobilized antibody- polypeptide complex for an amount of time sufficient to detect the bound antibody. An appropriate amount of time may generally be determined from the manufacturer's instructions or by assaying the level of binding that occurs over a period of time. Unbound detection reagent is then removed and bound detection reagent is detected using the reporter group.
  • the method employed for detecting the reporter group depends upon the nature of the reporter group. For radioactive groups, scintillation counting or autoradiographic methods are generally appropriate. Spectroscopic methods may be used to detect dyes, luminescent groups and fluorescent groups.
  • Biotin may be detected using avidin, coupled to a different reporter group (commonly a radioactive or fluorescent group or an enzyme). Enzyme reporter groups may generally be detected by the addition of substrate (generally for a specific period of time), followed by spectroscopic or other analysis of the reaction products.
  • reporter group commonly a radioactive or fluorescent group or an enzyme.
  • Enzyme reporter groups may generally be detected by the addition of substrate (generally for a specific period of time), followed by spectroscopic or other analysis of the reaction products.
  • the signal detected from the reporter group that remains bound to the solid support is generally compared to a signal that corresponds to a predetermined cut-off value.
  • This cut-off value is preferably the average mean signal obtained when the immobilized antigen is incubated with samples from an uninfected patient.
  • a sample generating a signal that is three standard deviations above the mean is considered positive for Leishmania antibodies and Leishmania infection.
  • the cut-off value is determined using a Receiver Operator Curve, according to the method of Sackett et al., Clinical Epidemiology: A Basic Science for Clinical Medicine, p. 106-7 (Little Brown and Co., 1985).
  • the cut-off value may be determined from a plot of pairs of true positive rates (i.e., sensitivity) and false positive rates (100%-specificity) that correspond to each possible cut-off value for the diagnostic test result.
  • the cut-off value on the plot that is the closest to the upper left-hand corner i.e., the value that encloses the largest area
  • a sample generating a signal that is higher than the cutoff value determined by this method may be considered positive.
  • the cutoff value may be shifted to the left along the plot, to minimize the false positive rate, or to the right, to minimize the false negative rate.
  • a sample generating a signal that is higher than the cut-off value determined by this method is considered positive for Leishmania infection.
  • a diagnostic assay may be performed in a flow- through or strip test format, wherein the antigen or fusion polypeptide is immobilized on a membrane such as nitrocellulose.
  • a detection reagent e.g., protein A-colloidal gold
  • a detection reagent then binds to the antibody-polypeptide complex as the solution containing the detection reagent flows through the membrane. The detection of bound detection reagent may then be performed as described above.
  • one end of the membrane to which polypeptide is bound is immersed in a solution containing the sample.
  • the sample migrates along the membrane through a region containing detection reagent and to the area of immobilized polypeptide. Concentration of detection reagent at the polypeptide indicates the presence of Leishmania antibodies in the sample.
  • Such tests can typically be performed with a very small amount (e.g., one drop) of patient serum or blood.
  • methods are provided for detecting
  • Leishmania in a biological sample using antibodies which may be polyclonal or monoclonal
  • T-cells specific for one or more antigens, fusion polypeptides and/or immunogenic portions of the invention are also useful as antibodies.
  • mice Female C57BL/6 mice were purchased from Charles River Laboratories (Wilmington, MA), and were maintained in specific-pathogen-free conditions. Eight to twelve-week-old mice at the beginning of experiments were used. Promastigotes of L. infantum (MHOM/BR/82/BA-2) were cultured in MEM (Invitrogen, Carlsbad, CA) supplemented with 0.5X MEM essential amino acids solution (Invitrogen), 0.1 mM MEM non-essential amino acids (Invitrogen), 1 mM sodium pyruvate, 25 mM HEPES, 8.3 mM glucose, 26 mM sodium bicarbonate, 1 ⁇ g/ml para amino benzoic acid, 50 ⁇ g/ml gentamicin 10% heat-inactivated fetal bovine serum and 6 ⁇ g/ml hemin.
  • MEM essential amino acids solution
  • Invitrogen 0.1 mM MEM non-essential amino acids
  • 1 mM sodium pyruvate 25 m
  • L. infantum SMT An open reading frame of L. infantum SMT was amplified by PCR using L. infantum genomic DNA with a set of primers, 5' CAA TTA CAT ATG TCC GCC GGT GGC CGT G (SEQ ID NO: 9), 3' CAA TTA AAG CTT CTA AGC CTG CTT GGA CGG (SEQ ID NO: 10).
  • the amplified PCR product was inserted in-frame with a 6xHis tag into the vector pET-28a (EMD Biosciences, San Diego, CA) and the insert was sequenced.
  • the deduced amino acid sequence of L. infantum SMT was compared with those of SMTs from L. donovani (Accession No. AAR92098), L.
  • CAJ09196 Trypanosoma cruzi (EAN81270) and Candida albicans (AAC26626), which were obtained from the NCBI database (www.ncbi.nlm.nih.gov), using the MegAlign software package (DNASTAR Inc., Madison, WI) by the Clustal method.
  • the pET-28a vector cloned for L. infantum SMT was transformed into E. coli Rosetta. Expression of the recombinant protein was induced by cultivation with IM isopropyl- ⁇ -D-thiogalactoside. rSMT was then purified as 6x His-tagged proteins using Ni-NTA agarose (Qiagen Inc., Valencia, CA).
  • mice were immunized with 10 ⁇ g of rSMT plus 20 ⁇ g of MPL ® -SE (GlaxoSmithKline Biologicals, Rixensant, Belgium) in a volume of 0.1 ml. Another group of mice was administrated with 10 ⁇ g of rSMT alone. Control groups received either saline or MPL ® -SE alone. The mice were immunized subcutaneously three times at three weeks intervals in the right hind footpad and at the base of the tail.
  • MPL ® -SE GaxoSmithKline Biologicals, Rixensant, Belgium
  • Samples for immunoblotting were prepared by suspending promastigotes in SDS sample buffer followed by boiling for 5 min. Samples containing 5 x 10 5 promastigotes were separated by SDS-PAGE and blotted on polyvinylidene difluoride membranes. Polyclonal Ab obtained from mice immunized with rSMT plus MPL -SE was used for western blotting. Sera from na ⁇ ve mice were used at the same dilution as a control. The membranes were then probed with HRP-conjugated goat anti-mouse IgG. (Rockland Immunochemicals, Inc., Gilbertsville, PA). Development was performed using Chemiluminescent Super Sensitive HRP Substrate Kit (BioFX Laboratories, Owings Mills, MD).
  • IgG subclass assay HRP- conjugated anti-human IgGl, IgG2, IgG3 or IgG4 (Invitrogen) were used.
  • the plates were developed with tetramethylbenzidine peroxidase substrate (Kirkegaard & Perry Laboratories, Gaithersburg, MD) and read by a microplate reader at 450 nm (570 nm reference).
  • the ELISA protocol was similar to that described above but different primary and secondary antibodies were used. Three mice per group were bled one week after the last immunization for Ab ELISA. Mouse serum samples were diluted to 1 :100 and applied to the plates in fivefold serial dilutions. Then the plates were incubated with HRP-conjugated goat anti-mouse IgGl or IgG2a (Southern Biotech, Birmingham, AL). The plates were developed with the substrate and read by a microplate reader at a 450 nm wavelength. Endpoint titers were calculated with the GraphPad Prism software using an OD value of 0.1 as a cutoff. 7. Cytokine assay using spleen cells
  • Spleens were collected from three mice per group two weeks after the last immunization. 2 x 10 5 splenocytes in complete RPMI medium (RPMI supplemented with 10% heat-inactivated fetal bovine serum, 100 U/ml of penicillin and 100 ⁇ g/ml of streptomycin) per well were plated in a 96-well plate and then stimulated with 3 ⁇ g/ml of con A, 100 ⁇ g/ml of L infantum soluble lysate antigen (LiSLA), 10 ⁇ g/ml of rSMT or medium alone. Culture supernatants were collected after 72 hrs cultivation and tested the levels of IFN- ⁇ and IL-10 by sandwich ELISA. 8.
  • Intracellular staining and Flow cytometry Spleens were collected from three mice per group two weeks after the last immunization.
  • 1 x 10 6 splenocytes in 100 ⁇ l of complete RPMI per well were plated in a round bottom 96-well plate and then stimulated with PMA/ionomycin, 10 ⁇ g/ml of rSMT or medium alone.
  • Co-stimulation antibodies anti-CD28 (eBioscience, San Diego, CA) and anti-CD49d (eBioscience) were added to the media for a final concentration of 1 ⁇ g/ml in the well during stimulation.
  • brefeldin A GolgiPlug: BD Biosciences, San Jose, CA
  • brefeldin A GolgiPlug: BD Biosciences, San Jose, CA
  • AlexaFluor 700-anti-CD3 eBioscience
  • PerCP-anti-CD4 BD Biosciences
  • PE-anti-CD8 BD Biosciences
  • mice per group were challenged with L. infantum three weeks after the last immunization.
  • 5 x 10 6 Z. infantum promastigotes were suspended in Hank's balanced salt solution and injected i.v. into the tail vein of the mouse.
  • mice were sacrificed to collect spleens and livers to determine the numbers of parasites in these tissues by limiting dilution assay.
  • the tissues were homogenated with glass grinders and the suspensions were twofold serially diluted with complete HOMEM in 96-well microplates with NNN blood agar. Each well was examined for the presence of parasites ten days after plating, and the numbers of parasites in the original tissues were calculated based on dilution factor of the last positive well.
  • rSMT was expressed in E. coli and purified (Fig. 2). A n apparent molecular mass of the protein was as predicted (42 kDa). Mouse polyclonal Ab raised against rSMT was used for detection of native SMT in Leishmania species by western blot analysis. Anti-SMT Ab detected a band with apparent molecular sizes of 38 kDa, which is in a same range of the predicted size, in all the Leishmania species tested, i.e. L. donovani, L infantum and L. major, whereas the intensity of the bands was different (Fig. 3). Those bands were not detected when sera from na ⁇ ve mice were used as the primary Ab.
  • VL patient sera recognize rSMT To determine antigenicity of Leishmania SMT in humans, examined the prevalence of antibodies to rSMT and rK39 in VL patients by ELISA using sera from 21 Brazilian VL patients. rK39 is a serodiagnostic antigen and the presence of antibodies to the antigen indicates active VL (Burns et al., (1993) Proc Natl Acad Sci U S A 90:775779). 19 of the 21 showed strong Ab responses to rK39 and those responses were specific in VL patients (Fig. 4A). Those sera showed moderate to strong reactivity to rSMT, and these Ab responses seemed to be specific in VL patients, whereas one non-endemic healthy donor showed moderate response (Fig. 4B).
  • IgG2 and IgG3 responses to rK39 were detected in VL patient sera and IgGl was the predominant subclass (Fig. 4C). IgGl response was predominant to rSMT as well, despite the titers were lower than those to rK39 (Fig. 4D). Some IgG2 responses were observed in VL patient sera to rSMT, but that was not VL-specific, as health donors showed IgG2 response to this antigen. Little IgG4 responses were detected to either rK39 or rSMT.
  • mice immunized with rSMT plus MPL ® -SE showed robust humoral responses to rSMT characterized by high levels of antigen-specific IgGl and IgG2a (Fig. 5).
  • immunization with rSMT alone resulted in IgGl -dominant Ab responses.
  • Mice injected with saline or adjuvant alone showed no Ab responses to rSMT.
  • cytokine production by spleen cells in stimulation with rSMT or LiSLA were measured.
  • Spleen cells from mice immunized with rSMT plus MPL -SE produced a high level of IFN- ⁇ in response to rSMT (Fig. 6A). These mice also responded to LiSLA stimulation; albeit the magnitude of IFN- ⁇ production was much lower than that observed with rSMT stimulation. In contrast, spleen cells from mice immunized with rSMT alone produced only a low level of IFN- ⁇ in response to rSMT. Compared with administration of SMT alone, IFN- ⁇ /IL-10 ratio was drastically improved when MPL ® -SE was co-injected with the antigen (Fig. 6B). No detectable cytokine production was found in saline or adjuvant alone groups in stimulation with rSMT or LiSLA.
  • rSMT plus MPL ® -SE induce both CD4 + and CD8 + cells expressing multiple ThI -type cytokines
  • FACS data show that antigen-specific CD4 + and CD8 + cells were induced by rSMT plus MPL ® -SE vaccination (Fig. 7B).
  • CD4 + cells producing TNF- ⁇ , IL-2 or IFN- ⁇ in stimulation with rSMT were found only in that group of mice.
  • Antigen-specific CD8 + cells were also found in mice administrated rSMT plus MPL ® - SE, and the frequency of those cells producing TNF- ⁇ or IFN- ⁇ was high.
  • CD4 + and CD8 + cells were further divided into seven distinct populations based on expression patterns of TNF- ⁇ , IL-2 or IFN- ⁇ at a single-cell level, it was revealed that many of antigen-specific T-cells producing multiple cytokines were induced by rSMT plus MPL ® -SE (Fig. 7C). There were both CD4 + and CD8 + cells producing only TNF- ⁇ in stimulation with rSMT, whereas cells expressing only IFN- ⁇ were little. Most of antigen-specific CD4 + T-cells producing IFN- ⁇ were co-expressing TNF- ⁇ or both TNF- ⁇ and IL-2. Also, many of the antigen-specific CD8 + T-cells were producing both TNF- ⁇ and IFN- ⁇ .
  • mice are resistant against L. infantum infection
  • immunized mice were challenged by intravenous injection of 5 x 10 6 I. infantum promastigotes.
  • I. infantum promastigotes.
  • C57BL/6 mice showed peak parasite burden in livers around four weeks of the infection (data not shown).
  • Significant reduction of parasites was seen in mice immunized with rSMT plus MPL ® -SE compared with those in saline or adjuvant alone groups (Fig. 8).
  • mice showed 43-fold and 55-fold reduction in the number of parasites in spleens, 111 -fold and 117-fold reduction in livers compared with saline and adjuvant alone groups, respectively. There was no significant difference observed between saline and adjuvant alone groups. This protection in SMT-immunized mice was repeatedly observed with similar magnitude.
  • SMT is expressed by both L. infantum and L. donovani, the two causative agents of VL, as well as L major, a causative agent of CL, and L. braziliensis, a causative agent of both CL and disseminated leishmaniasis (DL).
  • This SMT expression pattern suggests that the antigen may also be useful in other forms of leishmaniasis.
  • the lack of homology with mammalian proteins is expected to offer safety advantages when vaccinating with SMT. SMT is also found in other pathogens including parasites and fungi such as Trypanosoma spp.
  • SMT also represents a vaccine candidate for treating conditions other than VL.
  • mice (Charles River Laboratories) were maintained in specific- pathogen-free conditions. Mice were eight to twelve weeks old at the beginning of experiments.
  • Promastigotes of L. major (MHOM/IL/80/Friedlin) were grown at 25 0 C in medium 199 supplemented with 20% heat-inactivated fetal bovine serum, 100 U of penicillin per ml, 100 ⁇ g of streptomycin per ml, 2 mM L-glutamine, 0.1 mM adenine, 40 mM HEPES, 0.25 mg of hemin per ml, and 0.31 mg of 6-biotin per ml.
  • Promastigotes in a late log or stationary phase were used for infections or antigen preparations.
  • mice Groups of five mice were immunized. The first group was immunized with saline as a negative control. The second group was immunized with 10 ⁇ g of rSMT plus 20 ⁇ g of MPL ® -SE (GlaxoSmithKline Biologicals, Rixensant, Belgium) in a volume of 0.1 ml. As a challenge, 2,000 L. major promastigotes were suspended in 10 ⁇ l of phosphate buffered saline and injected intradermally into both the left and right ears. Mice were infected 3 weeks after completion of the immunization protocol.
  • mice immunized with rSMT were significantly protected against L. major, a causative agent of cutaneous leishmaniasis.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Mycology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Enzymes And Modification Thereof (AREA)

Abstract

L'invention concerne des compositions et des procédés pour prévenir, traiter et détecter des leishmanioses. Les compositions comprennent d'une manière générale des polypeptides, des parties, des variantes et/ou fusions de stérol 24-C-méthyltransférase (SMT) de leishmania, ainsi que des polynucléotides codant des polypeptides, des parties, des variantes et/ou fusions de SMT.
EP08781725A 2007-07-13 2008-07-11 Compositions de stérol 24-c-méthyltransférase de leishmania pour la prévention, le traitement et le diagnostic de leishmanioses Withdrawn EP2170372A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US94963707P 2007-07-13 2007-07-13
PCT/US2008/069843 WO2009012166A1 (fr) 2007-07-13 2008-07-11 Compositions de stérol 24-c-méthyltransférase de leishmania pour la prévention, le traitement et le diagnostic de leishmanioses

Publications (1)

Publication Number Publication Date
EP2170372A1 true EP2170372A1 (fr) 2010-04-07

Family

ID=39760688

Family Applications (1)

Application Number Title Priority Date Filing Date
EP08781725A Withdrawn EP2170372A1 (fr) 2007-07-13 2008-07-11 Compositions de stérol 24-c-méthyltransférase de leishmania pour la prévention, le traitement et le diagnostic de leishmanioses

Country Status (7)

Country Link
US (1) US8916168B2 (fr)
EP (1) EP2170372A1 (fr)
CN (1) CN101743016B (fr)
BR (1) BRPI0815565A2 (fr)
EA (1) EA017958B1 (fr)
HK (1) HK1144915A1 (fr)
WO (1) WO2009012166A1 (fr)

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7833534B2 (en) 2006-04-10 2010-11-16 Infectious Disease Research Institute Compounds and methods for diagnosis and treatment of leishmaniasis
US8410258B2 (en) 2008-05-21 2013-04-02 Infections Disease Research Institute Recombinant polyprotein vaccines for the treatment and diagnosis of leishmaniasis
AR073170A1 (es) * 2008-05-21 2010-10-20 Infectious Disease Res Inst Vacunas de poliproteinas recombinantes para el tratamiento y diagnostico de leishmaniasis
ES2628743T3 (es) 2008-07-03 2017-08-03 Infectious Disease Research Institute Proteínas de fusión y su uso en el diagnóstico y tratamiento de la leishmaniasis
ES2352780B2 (es) * 2009-06-04 2011-09-15 Laboratorios Ovejero S.A. Lipopolisacárido de ochrobactrum intermedium contra la sepsis.
SA110310855B1 (ar) * 2009-11-13 2014-09-16 Laboratories Leti S L Unipersonal استخدام مصدر l3 و/ أو l5 كلقاح أو كوسيلة تشخيصية لمرض طفيلي
WO2012064659A1 (fr) * 2010-11-08 2012-05-18 Infectious Disease Research Institute Vaccins comprenant des polypeptides d'hydrolase nucléosidique et de stérol 24-c-méthyltransférase (smt) non spécifiques destinés à traiter et à diagnostiquer la leishmaniose
BR112015024860B1 (pt) 2013-03-28 2023-11-07 Access To Advanced Health Institute Polipeptídeo de fusão, polinucleotídeo isolado, composição compreendendo dito polipeptídeo, uso da dita composição para estimular uma resposta imune contra leishmania e método in vitro e kit de diagnóstico para detectar infecção por leishmania em uma amostra biológica
US9504743B2 (en) 2013-09-25 2016-11-29 Sequoia Sciences, Inc Compositions of vaccines and adjuvants and methods for the treatment of urinary tract infections
US9149522B2 (en) 2013-09-25 2015-10-06 Sequoia Sciences, Inc. Compositions of vaccines and adjuvants and methods for the treatment of urinary tract infections
US9149521B2 (en) 2013-09-25 2015-10-06 Sequoia Sciences, Inc. Compositions of vaccines and adjuvants and methods for the treatment of urinary tract infections
US20150086592A1 (en) 2013-09-25 2015-03-26 Sequoia Sciences, Inc Compositions of vaccines and adjuvants and methods for the treatment of urinary tract infections
US11801223B2 (en) 2013-12-31 2023-10-31 Access To Advanced Health Institute Single vial vaccine formulations
MX2018014399A (es) 2016-06-01 2019-06-06 Infectious Disease Res Inst Particulas de nanoalumbre que contienen un agente de dimensionamiento.
CA3141577A1 (fr) 2019-05-25 2020-12-03 Infectious Disease Research Institute Composition et procede de sechage par atomisation d'une emulsion de vaccin et d'adjuvant
CN117120087A (zh) 2020-12-23 2023-11-24 高级健康研究所 茄尼醇疫苗助剂及其制备方法
CN114250206B (zh) * 2021-12-25 2024-03-19 苏州瀚源新酶生物科技有限公司 甲基转移酶突变体、重组载体、重组工程菌及其应用

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6500437B1 (en) * 1995-09-22 2002-12-31 Corixa Corporation Leishmania antigens for use in the therapy and diagnosis of leishmaniasis
US6613337B1 (en) * 1997-02-12 2003-09-02 Corixa Corporation Leishmania antigens for use in the therapy and diagnosis of leishmaniasis

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
None *
See also references of WO2009012166A1 *

Also Published As

Publication number Publication date
CN101743016B (zh) 2015-05-13
US8916168B2 (en) 2014-12-23
BRPI0815565A2 (pt) 2015-02-18
HK1144915A1 (en) 2011-03-18
EA201070140A1 (ru) 2010-08-30
WO2009012166A1 (fr) 2009-01-22
US20090041798A1 (en) 2009-02-12
EA017958B1 (ru) 2013-04-30
CN101743016A (zh) 2010-06-16

Similar Documents

Publication Publication Date Title
US8916168B2 (en) Leishmania sterol 24-c-methyltransferase compositions for the prevention, treatment and diagnosis of leishmaniasis
US8425919B2 (en) Recombinant polyprotein vaccines for the treatment and diagnosis of leishmaniasis
EP2270170B1 (fr) Composés pour l'immunothérapie et le diagnostic de la tuberculose et leur utilisation
CA2230885C (fr) Composes et methodes d'immunotherapie et de diagnostic de la tuberculose
US8410258B2 (en) Recombinant polyprotein vaccines for the treatment and diagnosis of leishmaniasis
EP2637687B1 (fr) Vaccins comprenant des polypeptides d'hydrolase nucléosidique et de stérol 24-c-méthyltransférase (smt) non spécifiques destinés à traiter et à diagnostiquer la leishmaniose
WO1997009428A9 (fr) Composes et methodes d'immunotherapie et de diagnostic de la tuberculose
US9909114B2 (en) Vaccines comprising leishmania polypeptides for the treatment and diagnosis of leishmaniasis
WO2018053294A1 (fr) Vaccins comprenant des polypeptides de mycobacterium leprae pour la prévention, le traitement et le diagnostic de la lèpre
US20160158329A1 (en) Vaccines comprising leishmania polypeptides for the treatment and diagnosis of leishmaniasis

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20100129

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA MK RS

DAX Request for extension of the european patent (deleted)
17Q First examination report despatched

Effective date: 20160616

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20170919

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20180130